Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Public Health Manag Pract ; 29(6): 845-853, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37738597

RESUMEN

CONTEXT: Prior to the COVID-19 pandemic, wastewater influent monitoring for tracking disease burden in sewered communities was not performed in Ohio, and this field was only on the periphery of the state academic research community. PROGRAM: Because of the urgency of the pandemic and extensive state-level support for this new technology to detect levels of community infection to aid in public health response, the Ohio Water Resources Center established relationships and support of various stakeholders. This enabled Ohio to develop a statewide wastewater SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) monitoring network in 2 months starting in July 2020. IMPLEMENTATION: The current Ohio Coronavirus Wastewater Monitoring Network (OCWMN) monitors more than 70 unique locations twice per week, and publicly available data are updated weekly on the public dashboard. EVALUATION: This article describes the process and decisions that were made during network initiation, the network progression, and data applications, which can inform ongoing and future pandemic response and wastewater monitoring. DISCUSSION: Overall, the OCWMN established wastewater monitoring infrastructure and provided a useful tool for public health professionals responding to the pandemic.


Asunto(s)
COVID-19 , Aguas Residuales , Humanos , Ohio , Pandemias/prevención & control , Salud Pública , COVID-19/epidemiología , COVID-19/prevención & control , SARS-CoV-2
3.
Sci Rep ; 11(1): 4495, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33627767

RESUMEN

The COVID-19 pandemic caused by the novel SARS-CoV-2 is more contagious than other coronaviruses and has higher rates of mortality than influenza. Identification of effective therapeutics is a crucial tool to treat those infected with SARS-CoV-2 and limit the spread of this novel disease globally. We deployed a bioinformatics workflow to identify candidate drugs for the treatment of COVID-19. Using an "omics" repository, the Library of Integrated Network-Based Cellular Signatures (LINCS), we simultaneously probed transcriptomic signatures of putative COVID-19 drugs and publicly available SARS-CoV-2 infected cell lines to identify novel therapeutics. We identified a shortlist of 20 candidate drugs: 8 are already under trial for the treatment of COVID-19, the remaining 12 have antiviral properties and 6 have antiviral efficacy against coronaviruses specifically, in vitro. All candidate drugs are either FDA approved or are under investigation. Our candidate drug findings are discordant with (i.e., reverse) SARS-CoV-2 transcriptome signatures generated in vitro, and a subset are also identified in transcriptome signatures generated from COVID-19 patient samples, like the MEK inhibitor selumetinib. Overall, our findings provide additional support for drugs that are already being explored as therapeutic agents for the treatment of COVID-19 and identify promising novel targets that are worthy of further investigation.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos/métodos , Antivirales/farmacología , COVID-19/genética , COVID-19/metabolismo , Biología Computacional/métodos , Bases de Datos Factuales , Descubrimiento de Drogas/métodos , Humanos , Pandemias , Preparaciones Farmacéuticas , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Transcriptoma/efectos de los fármacos
4.
Xenotransplantation ; 27(6): e12629, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32697003

RESUMEN

Progress has been made in overcoming antibody-mediated rejection of porcine xenografts by deleting pig genes that produce unique carbohydrate epitopes. Pigs deficient in galactose α-1,3 galactose (gene modified: GGTA1) and neu5Gc (gene modified: CMAH) have reduced levels of human antibody binding. Previously we identified α-fucose as a glycan that was expressed in high levels on cells of GGTA1/CMAH KO pigs. To validate the α-fucose phenotype observed previously we compared lectin affinity toward human and pig serum glycoproteins by dot blot analysis and confocal microscopy. Human anti-fucose antibody isolated by affinity chromatography was tested for specificity to L-fucose by custom macroarray. The affinity and cytotoxicity of the isolated human anti-fucose antibody toward human and GGTA1/CMAH KO pig PBMCs was determined by flow cytometry. Dot blot and confocal analysis support out previous findings that α-fucose is more highly expressed in pigs than humans. Pig kidney glomeruli and tubules contain abundant α-fucose and may represent focal sites for anti-α-fucose antibody binding. The Isolated human anti-fucose IgA, IgG and IgM bound to GGTA1/CMAH KO pig PBMC and were cytotoxic. Interestingly, the isolated human IgG cross reacted with the methyl pentose, L-rhamnose. Human anti-fucose antibody bound and was cytotoxic to GGTA1/CMAH KO pig peripheral blood monocytes. We have shown that α-fucose is an abundant target for cytotoxic human antibody in the organs of genetically modified pigs important to xenotransplantation.


Asunto(s)
Animales Modificados Genéticamente , Antígenos Heterófilos/inmunología , Fucosa , Trasplante Heterólogo , Animales , Fucosa/inmunología , Galactosiltransferasas , Técnicas de Inactivación de Genes , Humanos , Leucocitos Mononucleares , Oxigenasas de Función Mixta , Porcinos
5.
Res Sq ; 2020 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-32702077

RESUMEN

The COVID-19 pandemic caused by the novel SARS-CoV-2 is more contagious than other coronaviruses and has higher rates of mortality than influenza. As no vaccine or drugs are currently approved to specifically treat COVID-19, identification of effective therapeutics is crucial to treat the afflicted and limit disease spread. We deployed a bioinformatics workflow to identify candidate drugs for the treatment of COVID-19. Using an "omics" repository, the Library of Integrated Network-Based Cellular Signatures (LINCS), we simultaneously probed transcriptomic signatures of putative COVID-19 drugs and signatures of coronavirus-infected cell lines to identify therapeutics with concordant signatures and discordant signatures, respectively. Our findings include three FDA approved drugs that have established antiviral activity, including protein kinase inhibitors, providing a promising new category of candidates for COVID-19 interventions.

6.
J Biol Chem ; 295(20): 6811-6822, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32273341

RESUMEN

The interferon system is the first line of defense against virus infection. Recently, using a high-throughput genetic screen of a human interferon-stimulated gene short-hairpin RNA library, we identified a viral restriction factor, TDRD7 (Tudor domain-containing 7). TDRD7 inhibits the paramyxo-/pneumoviruses (e.g. Sendai virus and respiratory syncytial virus) by interfering with the virus-induced cellular autophagy pathway, which these viruses use for their replication. Here, we report that TDRD7 is a viral restriction factor against herpes simplex virus (HSV-1). Using knockdown, knockout, and ectopic expression systems, we demonstrate the anti-HSV-1 activity of TDRD7 in multiple human and mouse cell types. TDRD7 inhibited the virus-activated AMP-activated protein kinase (AMPK), which was essential for HSV-1 replication. Genetic ablation or chemical inhibition of AMPK activity suppressed HSV-1 replication in multiple human and mouse cells. Mechanistically, HSV-1 replication after viral entry depended on AMPK but not on its function in autophagy. The antiviral activity of TDRD7 depended on its ability to inhibit virus-activated AMPK. In summary, our results indicate that the newly identified viral restriction factor TDRD7 inhibits AMPK and thereby blocks HSV-1 replication independently of the autophagy pathway. These findings suggest that AMPK inhibition represents a potential strategy to manage HSV-1 infections.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Herpesvirus Humano 1/fisiología , Ribonucleoproteínas/metabolismo , Replicación Viral , Proteínas Quinasas Activadas por AMP/genética , Animales , Chlorocebus aethiops , Células HeLa , Humanos , Ratones , Ribonucleoproteínas/genética , Células Vero
7.
Viruses ; 12(4)2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32295140

RESUMEN

Interferon (IFN) regulatory factor 3 (IRF3) is the key transcription factor for the induction of IFN and antiviral genes. The absence of antiviral genes in IRF3 deficiency leads to susceptibility to a wide range of viral infections. Previously, we uncovered a function for nontranscriptional IRF3 (nt-IRF3), RLR (RIG-I-like receptor)-induced IRF3-mediated pathway of apoptosis (RIPA), which triggers apoptotic killing of virus-infected cells. Using knock-in mice expressing a transcriptionally inactive, but RIPA-active, IRF3 mutant, we demonstrated the relative contribution of RIPA to host antiviral defense. Given that RIPA is a cellular antiviral pathway, we hypothesized that small molecules that promote RIPA in virus-infected cells would act as antiviral agents. To test this, we conducted a high throughput screen of a library of FDA-approved drugs to identify novel RIPA activators. Our screen identified doxorubicin as a potent RIPA-activating agent. In support of our hypothesis, doxorubicin inhibited the replication of vesicular stomatitis virus, a model rhabdovirus, and its antiviral activity depended on its ability to activate IRF3 in RIPA. Surprisingly, doxorubicin inhibited the transcriptional activity of IRF3. The antiviral activity of doxorubicin was expanded to flavivirus and herpesvirus that also activate IRF3. Mechanistically, doxorubicin promoted RIPA by activating the extracellular signal-regulated kinase (ERK) signaling pathway. Finally, we validated these results using another RIPA-activating compound, pyrvinium pamoate, which showed a similar antiviral effect without affecting the transcriptional activity of IRF3. Therefore, we demonstrate that the RIPA branch of IRF3 can be targeted therapeutically to prevent virus infection.


Asunto(s)
Antivirales/farmacología , Apoptosis/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Factor 3 Regulador del Interferón/metabolismo , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Doxorrubicina/farmacología , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento/métodos , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Modelos Biológicos , Bibliotecas de Moléculas Pequeñas , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos
8.
Cell Rep ; 27(11): 3269-3283.e6, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31189110

RESUMEN

Tripartite motif-containing protein 5α (TRIM5α) is a cellular antiviral restriction factor that prevents early events in retrovirus replication. The activity of TRIM5α is thought to be limited to retroviruses as a result of highly specific interactions with capsid lattices. In contrast to this current understanding, we show that both human and rhesus macaque TRIM5α suppress replication of specific flaviviruses. Multiple viruses in the tick-borne encephalitis complex are sensitive to TRIM5α-dependent restriction, but mosquito-borne flaviviruses, including yellow fever, dengue, and Zika viruses, are resistant. TRIM5α suppresses replication by binding to the viral protease NS2B/3 to promote its K48-linked ubiquitination and proteasomal degradation. Importantly, TRIM5α contributes to the antiviral function of IFN-I against sensitive flaviviruses in human cells. Thus, TRIM5α possesses remarkable plasticity in the recognition of diverse virus families, with the potential to influence human susceptibility to emerging flaviviruses of global concern.


Asunto(s)
Infecciones por Flavivirus/metabolismo , Péptido Hidrolasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Animales , Factores de Restricción Antivirales , Gatos , Chlorocebus aethiops , Células Dendríticas/metabolismo , Células Dendríticas/virología , Flavivirus/patogenicidad , Flavivirus/fisiología , Infecciones por Flavivirus/virología , Células HEK293 , Humanos , Unión Proteica , Proteolisis , Especificidad por Sustrato , Ubiquitinación , Células Vero
9.
iScience ; 15: 489-501, 2019 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-31129244

RESUMEN

Tick-borne flaviviruses (TBFVs) can cause life-threatening encephalitis and hemorrhagic fever. To identify virus-host interactions that may be exploited as therapeutic targets, we analyzed the TBFV polyprotein in silico for antiviral protein-binding motifs. We obtained two putative tumor necrosis factor receptor-associated factor 6 (TRAF6)-binding motifs (TBMs) within the protease domain of the viral nonstructural 3 (NS3) protein. Here, we show that TBFV NS3 interacted with TRAF6 during infection and that TRAF6 supports TBFV replication. The proviral role of TRAF6 was not seen with mosquito-borne flaviviruses, consistent with the lack of conserved TBMs. Mutation of the second TBM within NS3 disrupted TRAF6 binding, coincident with reduced abundance of mature, autocatalytically derived form of the NS3 protease and significant virus attenuation in vitro. Our studies reveal insights into how flaviviruses exploit innate immunity for the purpose of viral replication and identify a potential target for therapeutic design.

11.
PLoS One ; 12(6): e0179781, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28650973

RESUMEN

Tick-borne flaviviruses (TBFVs), including Powassan virus and tick-borne encephalitis virus cause encephalitis or hemorrhagic fevers in humans with case-fatality rates ranging from 1-30%. Despite severe disease in humans, TBFV infection of natural rodent hosts has little noticeable effect. Currently, the basis for resistance to disease is not known. We hypothesize that the coevolution of flaviviruses with their respective hosts has shaped the evolution of potent antiviral factors that suppress virus replication and protect the host from lethal infection. In the current study, we compared virus infection between reservoir host cells and related susceptible species. Infection of primary fibroblasts from the white-footed mouse (Peromyscus leucopus, a representative host) with a panel of vector-borne flaviviruses showed up to a 10,000-fold reduction in virus titer compared to control Mus musculus cells. Replication of vesicular stomatitis virus was equivalent in P. leucopus and M. musculus cells suggesting that restriction was flavivirus-specific. Step-wise comparison of the virus infection cycle revealed a significant block to viral RNA replication, but not virus entry, in P. leucopus cells. To understand the role of the type I interferon (IFN) response in virus restriction, we knocked down signal transducer and activator of transcription 1 (STAT1) or the type I IFN receptor (IFNAR1) by RNA interference. Loss of IFNAR1 or STAT1 significantly relieved the block in virus replication in P. leucopus cells. The major IFN antagonist encoded by TBFV, nonstructural protein 5, was functional in P. leucopus cells, thus ruling out ineffective viral antagonism of the host IFN response. Collectively, this work demonstrates that the IFN response of P. leucopus imparts a strong and virus-specific barrier to flavivirus replication. Future identification of the IFN-stimulated genes responsible for virus restriction specifically in P. leucopus will yield mechanistic insight into efficient control of virus replication and may inform the development of antiviral therapeutics.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Virus de la Encefalitis Transmitidos por Garrapatas/patogenicidad , Interferón Tipo I/inmunología , Peromyscus/inmunología , Peromyscus/virología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Encefalitis Transmitida por Garrapatas/genética , Encefalitis Transmitida por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/virología , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferón Tipo I/antagonistas & inhibidores , Ratones , Peromyscus/genética , ARN Interferente Pequeño/genética , ARN Viral/genética , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteínas no Estructurales Virales/inmunología , Replicación Viral/genética , Replicación Viral/inmunología
12.
J Infect Dis ; 214(suppl 3): S355-S359, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27511894

RESUMEN

Antiviral therapeutics with existing clinical safety profiles would be highly desirable in an outbreak situation, such as the 2013-2016 emergence of Ebola virus (EBOV) in West Africa. Although, the World Health Organization declared the end of the outbreak early 2016, sporadic cases of EBOV infection have since been reported. Alisporivir is the most clinically advanced broad-spectrum antiviral that functions by targeting a host protein, cyclophilin A (CypA). A modest antiviral effect of alisporivir against contemporary (Makona) but not historical (Mayinga) EBOV strains was observed in tissue culture. However, this effect was not comparable to observations for an alisporivir-susceptible virus, the flavivirus tick-borne encephalitis virus. Thus, EBOV does not depend on (CypA) for replication, in contrast to many other viruses pathogenic to humans.


Asunto(s)
Antivirales/uso terapéutico , Ciclosporina/uso terapéutico , Brotes de Enfermedades , Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/tratamiento farmacológico , África Occidental/epidemiología , Fiebre Hemorrágica Ebola/virología , Humanos , Replicación Viral
13.
Cell Host Microbe ; 18(1): 61-74, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26159719

RESUMEN

Type I interferon (IFN-α/ß or IFN-I) signals through two receptor subunits, IFNAR1 and IFNAR2, to orchestrate sterile and infectious immunity. Cellular pathways that regulate IFNAR1 are often targeted by viruses to suppress the antiviral effects of IFN-I. Here we report that encephalitic flaviviruses, including tick-borne encephalitis virus and West Nile virus, antagonize IFN-I signaling by inhibiting IFNAR1 surface expression. Loss of IFNAR1 was associated with binding of the viral IFN-I antagonist, NS5, to prolidase (PEPD), a cellular dipeptidase implicated in primary immune deficiencies in humans. Prolidase was required for IFNAR1 maturation and accumulation, activation of IFNß-stimulated gene induction, and IFN-I-dependent viral control. Human fibroblasts derived from patients with genetic prolidase deficiency exhibited decreased IFNAR1 surface expression and reduced IFNß-stimulated signaling. Thus, by understanding flavivirus IFN-I antagonism, prolidase is revealed as a central regulator of IFN-I responses.


Asunto(s)
Dipeptidasas/metabolismo , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Interacciones Huésped-Patógeno , Interferón Tipo I/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal , Virus del Nilo Occidental/inmunología , Fibroblastos/inmunología , Humanos , Unión Proteica , Proteínas no Estructurales Virales/metabolismo
15.
Curr Opin Virol ; 9: 143-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25462446

RESUMEN

RNA viruses of plants and animals have polymerases that are error-prone and produce complex populations of related, but non-identical, genomes called quasispecies. While there are vast variations in mutation rates among these viruses, selection has optimized the exact error rate of each species to provide maximum speed of replication and amount of variation without losing the ability to replicate because of excessive mutation. High mutation rates result in the selection of populations increasingly robust, which means they are increasingly resistant to show phenotypic changes after mutation. It is possible to manipulate the mutation rate, either by the use of mutagens or by selection (or genetic manipulation) of fidelity mutants. These polymerases usually, but not always, perform as well as wild type (wt) during cell infection, but show major phenotypic changes during in vivo infection. Both high and low fidelity variants are attenuated when the wt virus is virulent in the host. Alternatively when wt infection is non-apparent, the variants show major restrictions to spread in the infected host. Manipulation of mutation rates may become a new strategy to develop attenuated vaccines for humans and animals.


Asunto(s)
Adaptación Biológica , Evolución Molecular , Mutación , Virus ARN/genética , Virus ARN/fisiología , Replicación Viral , Animales , Humanos , Tasa de Mutación , Plantas , Virus ARN/patogenicidad , Selección Genética
18.
J Gen Virol ; 94(Pt 2): 336-347, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23136362

RESUMEN

Toll-like receptor 7 (TLR7) recognizes guanidine-rich viral ssRNA and is an important mediator of peripheral immune responses to several ssRNA viruses. However, the role that TLR7 plays in regulating the innate immune response to ssRNA virus infections in specific organs such as the central nervous system (CNS) is not as clear. This study examined the influence of TLR7 on the neurovirulence of Langat virus (LGTV), a ssRNA tick-borne flavivirus. TLR7 deficiency did not substantially alter the onset or incidence of LGTV-induced clinical disease; however, it did significantly affect virus levels in the CNS with a log(10) increase in virus titres in brain tissue from TLR7-deficient mice. This difference in virus load was also observed following intracranial inoculation, indicating a direct effect of TLR7 deficiency on regulating virus replication in the brain. LGTV-induced type I interferon responses in the CNS were not dependent on TLR7, being higher in TLR7-deficient mice compared with wild-type controls. In contrast, induction of pro-inflammatory cytokines including tumour necrosis factor, CCL3, CCL4 and CXCL13 were dependent on TLR7. Thus, although TLR7 is not essential in controlling LGTV pathogenesis, it is important in controlling virus infection in neurons in the CNS, possibly by regulating neuroinflammatory responses.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Virus de la Encefalitis Transmitidos por Garrapatas/patogenicidad , Glicoproteínas de Membrana/inmunología , Neuronas/virología , Receptor Toll-Like 7/inmunología , Replicación Viral , Animales , Encéfalo/virología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis Transmitida por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/patología , Encefalitis Transmitida por Garrapatas/virología , Ratones , Ratones Noqueados , Carga Viral
19.
Cell Host Microbe ; 10(3): 185-96, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21925107

RESUMEN

In response to virus infection, type I interferons (IFNs) induce several genes, most of whose functions are largely unknown. Here, we show that the tripartite motif (TRIM) protein, TRIM79α, is an IFN-stimulated gene (ISG) product that specifically targets tick-borne encephalitis virus (TBEV), a Flavivirus that causes encephalitides in humans. TRIM79α restricts TBEV replication by mediating lysosome-dependent degradation of the flavivirus NS5 protein, an RNA-dependent RNA polymerase essential for virus replication. NS5 degradation was specific to tick-borne flaviviruses, as TRIM79α did not recognize NS5 from West Nile virus (WNV) or inhibit WNV replication. In the absence of TRIM79α, IFN-ß was less effective in inhibiting tick-borne flavivirus infection of mouse macrophages, highlighting the importance of a single virus-specific ISG in establishing an antiviral state. The specificity of TRIM79α for TBEV reveals a remarkable ability of the innate IFN response to discriminate between closely related flaviviruses.


Asunto(s)
Proteínas Portadoras/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Virus de la Encefalitis Transmitidos por Garrapatas/enzimología , Encefalitis Transmitida por Garrapatas/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Animales , Proteínas Portadoras/genética , Línea Celular , ARN Polimerasas Dirigidas por ADN/genética , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Virus de la Encefalitis Transmitidos por Garrapatas/fisiología , Encefalitis Transmitida por Garrapatas/genética , Encefalitis Transmitida por Garrapatas/virología , Humanos , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Proteínas Virales/genética
20.
Methods ; 55(2): 166-71, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21855635

RESUMEN

Ubiquitin (Ub) conjugation to a substrate protein is a widely used cellular mechanism for control of protein stability and function, modulation of signal transduction pathways and antiviral responses. Identification and characterization of ubiquitinated viral proteins is an important step in understanding novel mechanisms of viral protein regulation as well as elucidating cellular antiviral strategies. Here we describe a protocol to easily detect and characterize the ubiquitination status of a viral substrate protein expressed either during infection or ectopically expressed as a fusion with a biotinylatable epitope tag. This tag provides advantages over current immunoprecipitation techniques by making use of the extremely tight biotin-streptavidin interaction. We provide an example of this protocol using the nonstructural protein 5 (NS5) from Langat virus (LGTV), a member of the tick-borne encephalitis virus (TBEV) serocomplex within the Flavivirus genus. Using the protocols outlined here, we describe some of the pitfalls inherent in determination of Ub linkage and demonstrate that NS5 is modified by at least two distinct ubiquitination types, multiubiquitination and K48-linked polyubiquitin chains.


Asunto(s)
Proteínas Recombinantes de Fusión/metabolismo , Ubiquitina/metabolismo , Proteínas Ubiquitinadas/metabolismo , Ubiquitinación , Proteínas no Estructurales Virales/metabolismo , Sustitución de Aminoácidos , Virus de la Encefalitis Transmitidos por Garrapatas , Células HEK293 , Humanos , Inmunoprecipitación , Leupeptinas/farmacología , Inhibidores de Proteasoma , Proteínas Recombinantes de Fusión/aislamiento & purificación , Ubiquitina/genética , Proteínas Ubiquitinadas/aislamiento & purificación , Proteínas no Estructurales Virales/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...